Enabling immunotherapy for high-risk Group 3 medulloblastoma via systems immunology
通过系统免疫学对高危 3 组髓母细胞瘤进行免疫治疗
基本信息
- 批准号:10714138
- 负责人:
- 金额:$ 81.9万
- 依托单位:
- 依托单位国家:美国
- 项目类别:
- 财政年份:2023
- 资助国家:美国
- 起止时间:2023-09-07 至 2028-08-31
- 项目状态:未结题
- 来源:
- 关键词:AddressAdoptive Cell TransfersAtlasesBiologyBrain NeoplasmsCAR T cell therapyCD8-Positive T-LymphocytesCRISPR screenCancer EtiologyCell CommunicationCell TherapyCell physiologyCellsCellular biologyChildChildhood Malignant Brain TumorClassificationClustered Regularly Interspaced Short Palindromic RepeatsDataDevelopmentDiseaseExclusionExhibitsGeneticGenetically Engineered MouseGenomicsGoalsHeterogeneityHumanImmuneImmune EvasionImmunocompetentImmunologyImmunotherapeutic agentImmunotherapyJointsMacrophageMapsMediatingModelingMolecularMultiomic DataMusNetwork-basedPTPRC genePatientsPhenotypePre-Clinical ModelProteomicsRefractoryResistanceReverse engineeringSaint Jude Children&aposs Research HospitalSamplingSignal TransductionSortingSubgroupSystemSystems BiologyT cell responseT cell therapyT-LymphocyteTCF Transcription FactorTechnologyTestingTumor ImmunityTumor stageTumor-infiltrating immune cellsVisualizationcancer typechildhood cancer mortalitychimeric antigen receptor T cellsclinically relevantcloud baseddata visualizationdisorder riskexperimental studyfunctional genomicshigh riskhigh risk populationimprovedin vivoinsightmedulloblastomamouse modelmultiple omicsneoplastic cellnerve stem cellnotch proteinnoveloverexpressionscreeningsingle cell analysissingle-cell RNA sequencingspatiotemporalstem-like celltooltranscriptomic profilingtranscriptomicstumortumor microenvironmenttumor progressiontumor-immune system interactions
项目摘要
PROJECT SUMMARY / ABSTRACT
The goal of this project is to dissect the immune evasion mechanisms and enable immunotherapy for children
with high-risk Group 3 medulloblastoma (G3MB) via systems immunology approaches. Brain tumors are the
leading cause of cancer-related deaths in children. Medulloblastoma is the most prevalent malignant pediatric
brain tumor and is characterized by four major molecular subgroups, among which G3MB is the most aggressive
form and features MYC overexpression. The immunosuppressive tumor microenvironment (TME) is poorly
understood in G3MB, and no immunotherapy is available for children with this high-risk disease. Systems
immunology approaches—especially single-cell and spatial multi-omics profiling and in vivo CRISPR-based
functional screening—have proven powerful in dissecting tumor–TME interactions and identifying novel
immunotherapy targets in various cancer types, but very few studies have integrated these approaches. In our
preliminary studies, we applied two unique immunocompetent genetically-engineered mouse models (GEMMs)
of MYC-driven G3MB and performed scRNA-seq, scATAC-seq and spatial transcriptomics profiling. We enriched
immune cells from the TME by sorting CD45 positive cells for single-cell studies. Our preliminary analysis of
single-cell and spatial omics data revealed striking interactions of neural stem cell-like tumor cells with
macrophages and other immune cells that potentially create a suppressive TME and drive immune evasion in
mouse G3MB. We also performed in vivo CRISPR screening in tumor cells using the GEMMs to identify
modulators of tumor development, which demonstrated the feasibility of in vivo functional genomics screening
in our preclinical models. In this project, first, we propose to utilize cutting-edge single-cell and spatial omics
technologies to characterize the two G3MB GEMMs at different stages of tumor progression. We will use our
network-based tools to integrate these multi-omics data to dissect the dynamic tumor–immune interactions and
underlying “hidden” drivers that drive the immune exclusion and suppression during G3MB progression. We will
also validate discoveries of G3MB from mouse studies in patient samples. We will develop a cloud-based portal
to visualize and explore our single-cell and spatial data and tumor–TME interactomes of G3MB. Second, we will
establish the mechanistic basis of tumor–T cell interactions and strategies to enable adoptive T cell therapy for
G3MB by discovering functional drivers and putative targets in both tumor cells and T cells. To this end, we will
apply both candidate approach and in vivo CRISPR screening in immunocompetent GEMMs to identify tumor-
intrinsic modulators that will remodel the suppressive TME and sensitize G3MB tumors to adoptive T cell and
CAR-T cell therapies. We will also test if targeting inhibitory factors for T cell function will enable and optimize
effective adoptive T cell therapies against such tumors. Our studies promise to provide new insights into
mechanisms of tumor–TME interactions in G3MB and manifest legitimate immunotherapeutic opportunities.
项目摘要 /摘要
该项目的目的是剖析免疫避免机制,并为儿童提供免疫疗法
通过系统免疫学方法,具有高危组3髓母细胞瘤(G3MB)。脑肿瘤是
儿童与癌症相关死亡的主要原因。髓母细胞瘤是最普遍的恶性儿科
脑肿瘤,其特征是四个主要的分子亚组,其中G3MB是最具侵略性的
形式和特征MYC的过表达。免疫抑制性肿瘤微环境(TME)较差
在G3MB中了解齿,并且对于这种高风险疾病的儿童没有免疫疗法。系统
免疫学方法 - 尤其是单细胞和空间多词分析以及基于Vivo CRISPR的
功能性筛查 - 被证明在剖析肿瘤 - TME相互作用并识别新颖的方面被证明有力
各种癌症类型的免疫疗法靶标,但很少有研究整合了这些方法。在我们的
初步研究,我们应用了两个独特的免疫能力遗传工程小鼠模型(GEMM)
MYC驱动的G3MB并进行了SCRNA-SEQ,SCATAC-SEQ和空间转录组学分析。我们丰富了
通过对CD45阳性细胞排序单细胞研究,从TME中免疫细胞。我们对
单细胞和空间上的OMIC数据揭示了神经干细胞样肿瘤细胞与
巨噬细胞和其他可能产生抑制性TME并驱动免疫电池的免疫电池
鼠标G3MB。我们还使用宝石在肿瘤细胞中进行了体内CRISPR筛查以识别
肿瘤发育的调节剂,证明了体内功能基因组学筛查的可行性
在我们的临床前模型中。在这个项目中,首先,我们建议利用尖端的单细胞和空间OMIC
在肿瘤进展的不同阶段表征两个G3MB GEMM的技术。我们将使用我们的
基于网络的工具集成这些多摩管数据以剖析动态肿瘤 - 免疫相互作用和
在G3MB进展过程中驱动免疫排除和抑制的基础“隐藏”驱动程序。我们将
还从患者样品中的小鼠研究中验证了G3MB的发现。我们将开发基于云的门户
可视化和探索我们的单细胞和空间数据以及G3MB的肿瘤– TME相互作用。第二,我们会的
建立肿瘤– T细胞相互作用和策略的机械基础,以实现适应性T细胞治疗
通过在肿瘤细胞和T细胞中发现功能驱动因素和推定靶标,G3MB。为此,我们将
在免疫能力的宝石中同时应用候选方法和体内CRISPR筛查以鉴定肿瘤
内在调节剂将重塑抑制性TME和敏感的G3MB肿瘤以适应性T细胞和
CAR-T细胞疗法。我们还将测试针对T细胞功能的抑制因素是否将启用和优化
有效地针对此类肿瘤进行适应性T细胞疗法。我们的研究承诺将提供新的见解
G3MB中肿瘤与TME相互作用的机制和明显的合法免疫治疗机会。
项目成果
期刊论文数量(0)
专著数量(0)
科研奖励数量(0)
会议论文数量(0)
专利数量(0)
数据更新时间:{{ journalArticles.updateTime }}
{{
item.title }}
{{ item.translation_title }}
- DOI:
{{ item.doi }} - 发表时间:
{{ item.publish_year }} - 期刊:
- 影响因子:{{ item.factor }}
- 作者:
{{ item.authors }} - 通讯作者:
{{ item.author }}
数据更新时间:{{ journalArticles.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ monograph.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ sciAawards.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ conferencePapers.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ patent.updateTime }}
Hongbo Chi其他文献
Hongbo Chi的其他文献
{{
item.title }}
{{ item.translation_title }}
- DOI:
{{ item.doi }} - 发表时间:
{{ item.publish_year }} - 期刊:
- 影响因子:{{ item.factor }}
- 作者:
{{ item.authors }} - 通讯作者:
{{ item.author }}
{{ truncateString('Hongbo Chi', 18)}}的其他基金
Integrating systems immunology with immunometabolism and cancer immunity
将系统免疫学与免疫代谢和癌症免疫相结合
- 批准号:
10442703 - 财政年份:2021
- 资助金额:
$ 81.9万 - 项目类别:
Integrating systems immunology with immunometabolism and cancer immunity
将系统免疫学与免疫代谢和癌症免疫相结合
- 批准号:
10299800 - 财政年份:2021
- 资助金额:
$ 81.9万 - 项目类别:
2020 Immunometabolism in Health and Disease GRC
2020 健康与疾病中的免疫代谢 GRC
- 批准号:
9912281 - 财政年份:2021
- 资助金额:
$ 81.9万 - 项目类别:
Integrating systems immunology with immunometabolism and cancer immunity
将系统免疫学与免疫代谢和癌症免疫相结合
- 批准号:
10657475 - 财政年份:2021
- 资助金额:
$ 81.9万 - 项目类别:
Bidirectional metabolic signaling in follicular helper T cell differentiation
滤泡辅助 T 细胞分化中的双向代谢信号
- 批准号:
10687027 - 财政年份:2019
- 资助金额:
$ 81.9万 - 项目类别:
Bidirectional metabolic signaling in follicular helper T cell differentiation
滤泡辅助 T 细胞分化中的双向代谢信号
- 批准号:
10020901 - 财政年份:2019
- 资助金额:
$ 81.9万 - 项目类别:
Bidirectional metabolic signaling in follicular helper T cell differentiation
滤泡辅助 T 细胞分化中的双向代谢信号
- 批准号:
10466976 - 财政年份:2019
- 资助金额:
$ 81.9万 - 项目类别:
Bidirectional metabolic signaling in follicular helper T cell differentiation
滤泡辅助 T 细胞分化中的双向代谢信号
- 批准号:
10231172 - 财政年份:2019
- 资助金额:
$ 81.9万 - 项目类别:
Bidirectional metabolic signaling in follicular helper T cell differentiation
滤泡辅助 T 细胞分化中的双向代谢信号
- 批准号:
9917280 - 财政年份:2019
- 资助金额:
$ 81.9万 - 项目类别:
Regulation of TH17 plasticity and stemness by mTORC1
mTORC1 对 TH17 可塑性和干性的调节
- 批准号:
10208040 - 财政年份:2018
- 资助金额:
$ 81.9万 - 项目类别:
相似海外基金
Engineering T cells to overcome inhibitory receptor signals that limit the efficacy of adoptive cell therapy against ovarian cancer
改造 T 细胞以克服抑制性受体信号,这些信号限制了过继性细胞疗法对卵巢癌的疗效
- 批准号:
10526155 - 财政年份:2023
- 资助金额:
$ 81.9万 - 项目类别:
Time to ATTAC: Adoptive Transfer of T cells Against gp100+ Cells to treat LAM
ATTAC 时间:针对 gp100 细胞的 T 细胞过继转移来治疗 LAM
- 批准号:
10682121 - 财政年份:2023
- 资助金额:
$ 81.9万 - 项目类别:
Induction of autosis to overcome resistance in adoptive cell therapy for solid tumors
诱导自体死亡以克服实体瘤过继细胞治疗中的耐药性
- 批准号:
10629835 - 财政年份:2023
- 资助金额:
$ 81.9万 - 项目类别:
The role of epigenetic regulator UHRF1 in stability of induced regulatory T-cell function during influenza A virus-induced lung injury
表观遗传调节因子 UHRF1 在甲型流感病毒诱导的肺损伤过程中诱导调节 T 细胞功能稳定性中的作用
- 批准号:
10389878 - 财政年份:2023
- 资助金额:
$ 81.9万 - 项目类别:
Engineered TCR-Treg Cell Therapies Targeting Type 1 Diabetes Autoantigens
针对 1 型糖尿病自身抗原的工程化 TCR-Treg 细胞疗法
- 批准号:
10764143 - 财政年份:2023
- 资助金额:
$ 81.9万 - 项目类别: